Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Nat Commun ; 14(1): 7994, 2023 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-38042913

RESUMO

Aortic aneurysms, which may dissect or rupture acutely and be lethal, can be a part of multisystem disorders that have a heritable basis. We report four patients with deficiency of selenocysteine-containing proteins due to selenocysteine Insertion Sequence Binding Protein 2 (SECISBP2) mutations who show early-onset, progressive, aneurysmal dilatation of the ascending aorta due to cystic medial necrosis. Zebrafish and male mice with global or vascular smooth muscle cell (VSMC)-targeted disruption of Secisbp2 respectively show similar aortopathy. Aortas from patients and animal models exhibit raised cellular reactive oxygen species, oxidative DNA damage and VSMC apoptosis. Antioxidant exposure or chelation of iron prevents oxidative damage in patient's cells and aortopathy in the zebrafish model. Our observations suggest a key role for oxidative stress and cell death, including via ferroptosis, in mediating aortic degeneration.


Assuntos
Aneurisma Aórtico , Peixe-Zebra , Humanos , Masculino , Camundongos , Animais , Selenocisteína , Músculo Liso Vascular/metabolismo , Aneurisma Aórtico/genética , Aneurisma Aórtico/metabolismo , Selenoproteínas/genética , Miócitos de Músculo Liso/metabolismo
2.
Clin Endocrinol (Oxf) ; 97(4): 502-514, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35999191

RESUMO

Development and differentiation of the thyroid gland is directed by expression of specific transcription factors in the thyroid follicular cell which mediates hormone biosynthesis. Membrane transporters are rate-limiting for cellular entry of thyroid hormones (TH) (T4 and T3) into some tissues, with selenocysteine-containing, deiodinase enzymes (DIO1 and DIO2) converting T4 to the biologically active hormone T3. TH regulate expression of target genes via hormone-inducible nuclear receptors (TRα and TRß) to exert their physiological effects. Primary congenital hypothyroidism (CH) due to thyroid dysgenesis may be mediated by defects in thyroid transcription factors or impaired thyroid stimulating hormone receptor function. Dyshormonogenic CH is usually due to mutations in genes mediating thyroidal iodide transport, organification or iodotyrosine synthesis and recycling. Disorders of TH signalling encompass conditions due to defects in membrane TH transporters, impaired hormone metabolism due to deficiency of deiodinases and syndromes of Resistance to thyroid hormone due to pathogenic variants in either TRα or TRß. Here, we review the genetic basis, pathogenesis and clinical features of congenital, dysgenetic or dyshormonogenic hypothyroidism and disorders of TH transport, metabolism and action.


Assuntos
Hipotireoidismo , Hormônios Tireóideos , Humanos , Hipotireoidismo/genética , Iodeto Peroxidase/genética , Transdução de Sinais/genética , Hormônios Tireóideos/metabolismo , Fatores de Transcrição
3.
Mol Cell Biol ; 42(2): e0036321, 2022 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-34871063

RESUMO

Mutations in thyroid hormone receptor α (TRα), a ligand-inducible transcription factor, cause resistance to thyroid hormone α (RTHα). This disorder is characterized by tissue-specific hormone refractoriness and hypothyroidism due to the inhibition of target gene expression by mutant TRα-corepressor complexes. Using biophysical approaches, we show that RTHα-associated TRα mutants devoid of ligand-dependent transcription activation function unexpectedly retain the ability to bind thyroid hormone. Visualization of the ligand T3 within the crystal structure of a prototypic TRα mutant validates this notion. This finding prompted the synthesis of different thyroid hormone analogues, identifying a lead compound, ES08, which dissociates corepressor from mutant human TRα more efficaciously than T3. ES08 rescues developmental anomalies in a zebrafish model of RTHα and induces target gene expression in TRα mutation-containing cells from an RTHα patient more effectively than T3. Our observations provide proof of principle for developing synthetic ligands that can relieve transcriptional repression by the mutant TRα-corepressor complex for treatment of RTHα.


Assuntos
Proteínas Correpressoras/genética , Expressão Gênica/fisiologia , Predisposição Genética para Doença/genética , Hormônios Tireóideos/metabolismo , Animais , Humanos , Mutação/genética , Fenótipo , Receptores dos Hormônios Tireóideos/genética , Receptores alfa dos Hormônios Tireóideos/metabolismo , Tri-Iodotironina/genética
4.
Eur Thyroid J ; 10(6): 533-541, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34956926

RESUMO

INTRODUCTION: Resistance to thyroid hormone ß (RTHß) is an inherited syndrome caused by dominant negative variants in the THRB gene (NM_000461.5). The clinical picture of RTHß is variable, and patients harboring the same variant may display different degrees of disease severity. CASE PRESENTATION: A 30-year-old man presented with thyrotoxicosis and central hyperthyroidism and was found to have a novel variant in the exon 10 of THRB gene (c.C1282G, p.L428V), located within the third hot spot region of the C-terminal of the receptor. Surprisingly, the same variant was found in two other relatives with an apparent normal thyroid function at initial screening. After exclusion of a TSH-secreting adenoma and serum interference in the proband, and the finding that exogenous levothyroxine failed to suppress the TSH in the brother affected by nodular goiter, relatives' thyroid function tests (TFTs) were reassessed with additional analytical method revealing biochemical features consistent with RTHß in all carriers of the p.L428V variant. Functional studies showed a slightly impaired in vitro transcriptional activity of p.L428V. Interestingly' the expression of the human p.L428V thyroid hormone receptor beta in the zebrafish embryo background generated a phenotype consistent with RTHß. CONCLUSION: Variable results of TFTs on some immunoassays can be a cause of RTHß diagnostic delay, but the genotype-phenotype correlation in this family and functional studies support p.L428V as a novel THRB variant expanding the spectrum of gene variants causing RTHß. In vivo, rather than in vitro, functional assays may be required to demonstrate the dominant negative action of THRB variants.

5.
Diabetes ; 67(6): 1086-1092, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29622583

RESUMO

Loss-of-function mutations in PPARG cause familial partial lipodystrophy type 3 (FPLD3) and severe metabolic disease in many patients. Missense mutations in PPARG are present in ∼1 in 500 people. Although mutations are often binarily classified as benign or deleterious, prospective functional classification of all missense PPARG variants suggests that their impact is graded. Furthermore, in testing novel mutations with both prototypic endogenous (e.g., prostaglandin J2 [PGJ2]) and synthetic ligands (thiazolidinediones, tyrosine agonists), we observed that synthetic agonists selectively rescue function of some peroxisome proliferator-activated receptor-γ (PPARγ) mutants. We report on patients with FPLD3 who harbor two such PPARγ mutations (R308P and A261E). Both PPARγ mutants exhibit negligible constitutive or PGJ2-induced transcriptional activity but respond readily to synthetic agonists in vitro, with structural modeling providing a basis for such differential ligand-dependent responsiveness. Concordant with this finding, dramatic clinical improvement was seen after pioglitazone treatment of a patient with R308P mutant PPARγ. A patient with A261E mutant PPARγ also responded beneficially to rosiglitazone, although cardiomyopathy precluded prolonged thiazolidinedione use. These observations indicate that detailed structural and functional classification can be used to inform therapeutic decisions in patients with PPARG mutations.


Assuntos
Hipoglicemiantes/uso terapêutico , Lipodistrofia Parcial Familiar/tratamento farmacológico , Lipodistrofia Parcial Familiar/genética , Modelos Moleculares , Mutação de Sentido Incorreto , PPAR gama/genética , Tiazolidinedionas/uso terapêutico , Adolescente , Adulto , Substituição de Aminoácidos , Sítios de Ligação , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Genes Reporter/efeitos dos fármacos , Células HEK293 , Humanos , Hipoglicemiantes/efeitos adversos , Hipoglicemiantes/química , Hipoglicemiantes/farmacologia , Ligantes , Lipodistrofia Parcial Familiar/metabolismo , Conformação Molecular , Simulação de Acoplamento Molecular , PPAR gama/agonistas , PPAR gama/química , PPAR gama/metabolismo , Farmacogenética/métodos , Pioglitazona , Conformação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Rosiglitazona , Tiazolidinedionas/efeitos adversos , Tiazolidinedionas/química , Tiazolidinedionas/farmacologia , Adulto Jovem
6.
J Clin Endocrinol Metab ; 102(9): 3517-3525, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28911146

RESUMO

Context: Patients with resistance to thyroid hormone (TH) α (RTHα) are characterized by growth retardation, macrocephaly, constipation, and abnormal thyroid function tests. In addition, almost all RTHα patients have mild anemia, the pathogenesis of which is unknown. Animal studies suggest an important role for TH and TH receptor (TR)α in erythropoiesis. Objective: To investigate whether a defect in TRα affects the maturation of red blood cells in RTHα patients. Design, Setting, and Patients: Cultures of primary human erythroid progenitor cells (HEPs), from peripheral blood of RTHα patients (n = 11) harboring different inactivating mutations in TRα (P398R, F397fs406X, C392X, R384H, A382fs388X, A263V, A263S), were compared with healthy controls (n = 11). During differentiation, erythroid cells become smaller, accumulate hemoglobin, and express different cell surface markers. We assessed cell number and cell size, and used cell staining and fluorescence-activated cell sorter analysis to monitor maturation at different time points. Results: After ∼14 days of ex vivo expansion, both control and patient-derived progenitors differentiated spontaneously. However, RTHα-derived cells differentiated more slowly. During spontaneous differentiation, RTHα-derived HEPs were larger, more positive for c-Kit (a proliferation marker), and less positive for glycophorin A (a differentiation marker). The degree of abnormal spontaneous maturation of RTHα-derived progenitors did not correlate with severity of underlying TRα defect. Both control and RTHα-derived progenitors responded similarly when differentiation was induced. T3 exposure accelerated differentiation of both control- and RTHα patient-derived HEPs. Conclusions: Inactivating mutations in human TRα affect the balance between proliferation and differentiation of progenitor cells during erythropoiesis, which may contribute to the mild anemia seen in most RTHα patients.


Assuntos
Anemia/genética , Eritropoese/genética , Regulação da Expressão Gênica , Receptores alfa dos Hormônios Tireóideos/genética , Síndrome da Resistência aos Hormônios Tireóideos/genética , Adolescente , Adulto , Anemia/epidemiologia , Anemia/fisiopatologia , Estudos de Casos e Controles , Células Cultivadas , Criança , Pré-Escolar , Eritrócitos/metabolismo , Feminino , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , Mutação , Prognóstico , Valores de Referência , Papel (figurativo) , Células-Tronco/citologia , Células-Tronco/fisiologia , Síndrome da Resistência aos Hormônios Tireóideos/epidemiologia , Síndrome da Resistência aos Hormônios Tireóideos/fisiopatologia , Adulto Jovem
7.
Thyroid ; 27(7): 973-982, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28471274

RESUMO

BACKGROUND: Resistance to thyroid hormone alpha (RTHα), a disorder characterized by tissue-selective hypothyroidism and near-normal thyroid function tests due to thyroid receptor alpha gene mutations, is rare but probably under-recognized. This study sought to correlate the clinical characteristics and response to thyroxine (T4) therapy in two adolescent RTHα patients with the properties of the THRA mutation, affecting both TRα1 and TRα2 proteins, they harbored. METHODS: Clinical, auxological, biochemical, and physiological parameters were assessed in each patient at baseline and after T4 therapy. RESULTS: Heterozygous THRA mutations occurring de novo were identified in a 17-year-old male (patient P1; c.788C>T, p.A263V mutation) investigated for mild pubertal delay and in a 15-year-old male (patient P2; c.821T>C, p.L274P mutation) with short stature (0.4th centile), skeletal dysplasia, dysmorphic facies, and global developmental delay. Both individuals exhibited macrocephaly, delayed dentition, and constipation, together with a subnormal T4/triiodothyronine (T3) ratio, low reverse T3 levels, and mild anemia. When studied in vitro, A263V mutant TRα1 was transcriptionally impaired and inhibited the function of its wild-type counterpart at low (0.01-10 nM) T3 levels, with higher T3 concentrations (100 nM-1 µM) reversing dysfunction and such dominant negative inhibition. In contrast, L274P mutant TRα1 was transcriptionally inert, exerting significant dominant negative activity, only overcome with 10 µM of T3. Mirroring this, normal expression of KLF9, a TH-responsive target gene, was achieved in A263V mutation-containing peripheral blood mononuclear cells following 1 µM of T3 exposure, but with markedly reduced expression levels in L274P mutation-containing peripheral blood mononuclear cells, even with 10 µM of T3. Following T4 therapy, growth, body composition, dyspraxia, and constipation improved in P1, whereas growth retardation and constipation in P2 were unchanged. Neither A263V nor L274P mutations exhibited gain or loss of function in the TRα2 background, and no additional phenotype attributable to this was discerned. CONCLUSIONS: This study correlates a milder clinical phenotype and favorable response to T4 therapy in a RTHα patient (P1) with heterozygosity for mutant TRα1 exhibiting partial, T3-reversible, loss of function. In contrast, a more severe clinical phenotype refractory to hormone therapy was evident in another case (P2) associated with severe, virtually irreversible, dysfunction of mutant TRα1.


Assuntos
Mutação , Receptores alfa dos Hormônios Tireóideos/genética , Síndrome da Resistência aos Hormônios Tireóideos/genética , Tiroxina/uso terapêutico , Adolescente , Predisposição Genética para Doença , Genótipo , Humanos , Masculino , Fenótipo , Avaliação de Sintomas , Síndrome da Resistência aos Hormônios Tireóideos/diagnóstico , Síndrome da Resistência aos Hormônios Tireóideos/tratamento farmacológico , Resultado do Tratamento , Adulto Jovem
8.
Nat Genet ; 48(12): 1570-1575, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27749844

RESUMO

Clinical exome sequencing routinely identifies missense variants in disease-related genes, but functional characterization is rarely undertaken, leading to diagnostic uncertainty. For example, mutations in PPARG cause Mendelian lipodystrophy and increase risk of type 2 diabetes (T2D). Although approximately 1 in 500 people harbor missense variants in PPARG, most are of unknown consequence. To prospectively characterize PPARγ variants, we used highly parallel oligonucleotide synthesis to construct a library encoding all 9,595 possible single-amino acid substitutions. We developed a pooled functional assay in human macrophages, experimentally evaluated all protein variants, and used the experimental data to train a variant classifier by supervised machine learning. When applied to 55 new missense variants identified in population-based and clinical sequencing, the classifier annotated 6 variants as pathogenic; these were subsequently validated by single-variant assays. Saturation mutagenesis and prospective experimental characterization can support immediate diagnostic interpretation of newly discovered missense variants in disease-related genes.


Assuntos
Diabetes Mellitus Tipo 2/genética , Lipodistrofia/genética , Mutação de Sentido Incorreto/genética , Infarto do Miocárdio/genética , PPAR gama/genética , Substituição de Aminoácidos , Estudos de Casos e Controles , Feminino , Humanos , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Estudos Prospectivos
9.
J Clin Invest ; 126(3): 992-6, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26854926

RESUMO

Selenium is a trace element that is essential for human health and is incorporated into more than 25 human selenocysteine-containing (Sec-containing) proteins via unique Sec-insertion machinery that includes a specific, nuclear genome-encoded, transfer RNA (tRNA[Ser]Sec). Here, we have identified a human tRNA[Ser]Sec mutation in a proband who presented with a variety of symptoms, including abdominal pain, fatigue, muscle weakness, and low plasma levels of selenium. This mutation resulted in a marked reduction in expression of stress-related, but not housekeeping, selenoproteins. Evaluation of primary cells from the homozygous proband and a heterozygous parent indicated that the observed deficit in stress-related selenoprotein production is likely mediated by reduced expression and diminished 2'-O-methylribosylation at uridine 34 in mutant tRNA[Ser]Sec. Moreover, this methylribosylation defect was restored by cellular complementation with normal tRNA[Ser]Sec. This study identifies a tRNA mutation that selectively impairs synthesis of stress-related selenoproteins and demonstrates the importance of tRNA modification for normal selenoprotein synthesis.


Assuntos
Doenças Genéticas Inatas/diagnóstico , RNA de Transferência Aminoácido-Específico/genética , Selenoproteínas/genética , Sequência de Bases , Criança , Análise Mutacional de DNA , Estudos de Associação Genética , Doenças Genéticas Inatas/genética , Humanos , Masculino , Dados de Sequência Molecular , Mutação Puntual , Polimorfismo de Nucleotídeo Único , Biossíntese de Proteínas , Selenoproteínas/sangue , Selenoproteínas/deficiência
10.
Mol Cell Endocrinol ; 424: 102-17, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26802880

RESUMO

Resistance to thyroid hormone can be due to heterozygous, dominant negative (DN) THRA (RTHα) or THRB (RTHß) mutations, but the underlying mechanisms are incompletely understood. Here, we delineate the spatiotemporal expression of TH receptors (TRs) in zebrafish and generated morphants expressing equivalent amounts of wild-type and DN TRαs (thraa_MOs) and TRßs (thrb_MOs) in vivo. Both morphants show severe developmental abnormalities. The phenotype of thraa_MOs includes brain and cardiac defects, but normal thyroid volume and tshba expression. A combined modification of dio2 and dio3 expression can explain the high T3/T4 ratio seen in thraa_MOs, as in RTHα. Thrb_MOs show abnormal eyes and otoliths, with a typical RTHß pattern of thyroid axis. The coexpression of wild-type, but not mutant, human TRs can rescue the phenotype in both morphants. High T3 doses can partially revert the dominant negative action of mutant TRs in morphant fish. Therefore, our morphants recapitulate the RTHα and RTHß key manifestations representing new models in which the functional consequences of human TR mutations can be rapidly and faithfully evaluated.


Assuntos
Modelos Animais de Doenças , Receptores alfa dos Hormônios Tireóideos/genética , Receptores beta dos Hormônios Tireóideos/genética , Síndrome da Resistência aos Hormônios Tireóideos/genética , Hormônios Tireóideos/metabolismo , Peixe-Zebra/crescimento & desenvolvimento , Animais , Embrião não Mamífero , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Mutação , Receptores alfa dos Hormônios Tireóideos/metabolismo , Receptores beta dos Hormônios Tireóideos/metabolismo , Distribuição Tecidual , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
11.
Lancet Diabetes Endocrinol ; 2(8): 619-26, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24969835

RESUMO

BACKGROUND: The thyroid hormone receptor α gene (THRA) transcript is alternatively spliced to generate either thyroid hormone receptor (TR)α1 or a non-hormone-binding variant protein, TRα2, the function of which is unknown. Here, we describe the first patients identified with a mutation in THRA that affects both TRα1 and TRα2, and compare them with patients who have resistance to thyroid hormone owing to a mutation affecting only TRα1, to delineate the relative roles of TRα1 and TRα2. METHODS: We did clinical, biochemical, and genetic analyses of an index case and her two sons. We assessed physical and radiological features, thyroid function, physiological and biochemical markers of thyroid hormone action, and THRA sequence. FINDINGS: The patients presented in childhood with growth failure, developmental delay, and constipation, which improved after treatment with thyroxine, despite normal concentrations of circulating thyroid hormones. They had similar clinical (macrocephaly, broad faces, skin tags, motor dyspraxia, slow speech), biochemical (subnormal ratio of free thyroxine:free tri-iodothyronine [T3], low concentration of total reverse T3, high concentration of creatine kinase, mild anaemia), and radiological (thickened calvarium) features to patients with TRα1-mediated resistance to thyroid hormone, although our patients had a heterozygous mis-sense mutation (Ala263Val) in both TRα1 and TRα2 proteins. The Ala263Val mutant TRα1 inhibited the transcriptional function of normal receptor in a dominant-negative fashion. By contrast, function of Ala263Val mutant TRα2 matched its normal counterpart. In vitro, high concentrations of T3 restored transcriptional activity of Ala263Val mutant TRα1, and reversed the dominant-negative inhibition of its normal counterpart. High concentrations of T3 restored expression of thyroid hormone-responsive target genes in patient-derived blood cells. INTERPRETATION: TRα1 seems to be the principal functional product of the THRA gene. Thyroxine treatment alleviates hormone resistance in patients with mutations affecting this gene, possibly ameliorating the phenotype. These findings will help the diagnosis and treatment of other patients with resistance to thyroid hormone resulting from mutations in THRA. FUNDING: Wellcome Trust, NIHR Cambridge Biomedical Research Centre, Marie Curie Actions, Foundation for Development of Internal Medicine in Europe.


Assuntos
Processamento Alternativo , Mutação de Sentido Incorreto , Receptores alfa dos Hormônios Tireóideos/genética , Síndrome da Resistência aos Hormônios Tireóideos/genética , Adulto , Substituição de Aminoácidos , Saúde da Família , Feminino , Apraxia da Marcha/etiologia , Heterozigoto , Humanos , Masculino , Megalencefalia/etiologia , Pessoa de Meia-Idade , Pólipos/etiologia , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Neoplasias Cutâneas/etiologia , Distúrbios da Fala/etiologia , Receptores alfa dos Hormônios Tireóideos/agonistas , Receptores alfa dos Hormônios Tireóideos/metabolismo , Síndrome da Resistência aos Hormônios Tireóideos/tratamento farmacológico , Síndrome da Resistência aos Hormônios Tireóideos/fisiopatologia , Tiroxina/uso terapêutico , Resultado do Tratamento
12.
J Clin Endocrinol Metab ; 99(7): E1381-6, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24646103

RESUMO

CONTEXT: Familial dysalbuminemic hyperthyroxinemia, characterized by abnormal circulating albumin with increased T4 affinity, causes artefactual elevation of free T4 concentrations in euthyroid individuals. OBJECTIVE: Four unrelated index cases with discordant thyroid function tests in different assay platforms were investigated. DESIGN AND RESULTS: Laboratory biochemical assessment, radiolabeled T4 binding studies, and ALB sequencing were undertaken. (125)I-T4 binding to both serum and albumin in affected individuals was markedly increased, comparable with known familial dysalbuminemic hyperthyroxinemia cases. Sequencing showed heterozygosity for a novel ALB mutation (arginine to isoleucine at codon 222, R222I) in all four cases and segregation of the genetic defect with abnormal biochemical phenotype in one family. Molecular modeling indicates that arginine 222 is located within a high-affinity T4 binding site in albumin, with substitution by isoleucine, which has a smaller side chain predicted to reduce steric hindrance, thereby facilitating T4 and rT3 binding. When tested in current immunoassays, serum free T4 values from R222I heterozygotes were more measurably abnormal in one-step vs two-step assay architectures. Total rT3 measurements were also abnormally elevated. CONCLUSIONS: A novel mutation (R222I) in the ALB gene mediates dominantly inherited dysalbuminemic hyperthyroxinemia. Susceptibility of current free T4 immunoassays to interference by this mutant albumin suggests likely future identification of individuals with this variant binding protein.


Assuntos
Hipertireoxinemia Disalbuminêmica Familiar/genética , Mutação de Sentido Incorreto , Pré-Albumina/genética , Adulto , Substituição de Aminoácidos , Arginina/genética , Pré-Escolar , Feminino , Humanos , Hipertireoxinemia Disalbuminêmica Familiar/sangue , Isoleucina/genética , Masculino , Modelos Moleculares , Pré-Albumina/química , Testes de Função Tireóidea , Adulto Jovem
13.
J Clin Endocrinol Metab ; 98(11): 4254-61, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23940126

RESUMO

CONTEXT: The first human cases (female, age 6 y; father and daughter, ages 47 and 11 y, respectively) with growth retardation/short stature, skeletal dysplasia, constipation, and defective thyroid receptor α (TRα) have been recently described. OBJECTIVE: A 45-year-old, short, overweight female with cognitive impairment, epilepsy, and constipation was investigated. DESIGN AND INTERVENTION: Clinical, biochemical, and radiological assessment and THRA sequencing were undertaken. The patient's thyroid status and her biochemical and physiological parameters were evaluated at baseline and after T4 therapy. RESULTS: The patient exhibits disproportionate short stature, macrocephaly, low free T4/free T3 ratio and rT3 levels, together with subnormal heart and basal metabolic rate. She is heterozygous for a novel frameshift/premature stop (Ala382ProfsX7) THRA mutation, generating a mutant TRα with constitutive corepressor binding and negligible coactivator recruitment, which inhibits its wild-type counterpart in a dominant-negative manner-both in vitro and in mutation-containing patient blood mononuclear cells studied ex vivo. Her alertness and constipation responded to T4 therapy, which readily suppressed TSH levels, raised basal metabolic rate, and normalized elevated muscle creatine kinase, but cardiac parameters (heart rate, contractility) remained relatively refractory. The patient and a previous childhood case showed reduced red cell mass with macrocytosis unresponsive to T4 therapy. CONCLUSIONS: Clinical (short stature, macrocephaly, constipation) and biochemical (low free T4/free T3 ratio, subnormal rT3) findings that are congruent with previous cases and newly recognized features (epilepsy) in this adult female with defective TRα define a shared phenotype in TRα-mediated resistance to thyroid hormone, with differential tissue responses to T4 treatment.


Assuntos
Resistência a Medicamentos/genética , Hipotireoidismo/tratamento farmacológico , Hipotireoidismo/genética , Receptores alfa dos Hormônios Tireóideos/genética , Tiroxina/uso terapêutico , Saúde da Família , Feminino , Humanos , Hipotireoidismo/metabolismo , Pessoa de Meia-Idade , Receptores alfa dos Hormônios Tireóideos/metabolismo , Tiroxina/metabolismo
14.
N Engl J Med ; 366(3): 243-9, 2012 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-22168587

RESUMO

Thyroid hormones exert their effects through alpha (TRα1) and beta (TRß1 and TRß2) receptors. Here we describe a child with classic features of hypothyroidism (growth retardation, developmental retardation, skeletal dysplasia, and severe constipation) but only borderline-abnormal thyroid hormone levels. Using whole-exome sequencing, we identified a de novo heterozygous nonsense mutation in a gene encoding thyroid hormone receptor alpha (THRA) and generating a mutant protein that inhibits wild-type receptor action in a dominant negative manner. Our observations are consistent with defective human TRα-mediated thyroid hormone resistance and substantiate the concept of hormone action through distinct receptor subtypes in different target tissues.


Assuntos
Códon sem Sentido , Transtornos do Crescimento/genética , Hipotireoidismo/genética , Receptores alfa dos Hormônios Tireóideos/genética , Tiroxina/sangue , Tiroxina/uso terapêutico , Tri-Iodotironina/sangue , Criança , Feminino , Transtornos do Crescimento/tratamento farmacológico , Heterozigoto , Humanos , Hipotireoidismo/tratamento farmacológico , Modelos Moleculares , Conformação Proteica , Receptores alfa dos Hormônios Tireóideos/química , Hormônios Tireóideos/sangue
15.
J Clin Invest ; 120(12): 4220-35, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21084748

RESUMO

Selenium, a trace element that is fundamental to human health, is incorporated into some proteins as selenocysteine (Sec), generating a family of selenoproteins. Sec incorporation is mediated by a multiprotein complex that includes Sec insertion sequence-binding protein 2 (SECISBP2; also known as SBP2). Here, we describe subjects with compound heterozygous defects in the SECISBP2 gene. These individuals have reduced synthesis of most of the 25 known human selenoproteins, resulting in a complex phenotype. Azoospermia, with failure of the latter stages of spermatogenesis, was associated with a lack of testis-enriched selenoproteins. An axial muscular dystrophy was also present, with features similar to myopathies caused by mutations in selenoprotein N (SEPN1). Cutaneous deficiencies of antioxidant selenoenzymes, increased cellular ROS, and susceptibility to ultraviolet radiation-induced oxidative damage may mediate the observed photosensitivity. Reduced levels of selenoproteins in peripheral blood cells were associated with impaired T lymphocyte proliferation, abnormal mononuclear cell cytokine secretion, and telomere shortening. Paradoxically, raised ROS in affected subjects was associated with enhanced systemic and cellular insulin sensitivity, similar to findings in mice lacking the antioxidant selenoenzyme glutathione peroxidase 1 (GPx1). Thus, mutation of SECISBP2 is associated with a multisystem disorder with defective biosynthesis of many selenoproteins, highlighting their role in diverse biological processes.


Assuntos
Mutação , Proteínas de Ligação a RNA/genética , Selenoproteínas/deficiência , Adulto , Idoso , Sequência de Aminoácidos , Animais , Azoospermia/genética , Sequência de Bases , Criança , Pré-Escolar , Códon sem Sentido , DNA/genética , Feminino , Perda Auditiva Neurossensorial/genética , Humanos , Resistência à Insulina/genética , Masculino , Camundongos , Pessoa de Meia-Idade , Modelos Moleculares , Dados de Sequência Molecular , Distrofias Musculares/genética , Mutação de Sentido Incorreto , Linhagem , Transtornos de Fotossensibilidade/genética , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Selenocisteína/metabolismo , Selenoproteínas/metabolismo , Homologia de Sequência de Aminoácidos , Espermatogênese/genética , Linfócitos T/imunologia
16.
J Clin Endocrinol Metab ; 95(8): 4031-6, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20484477

RESUMO

CONTEXT: Homozygous loss-of-function mutations in forkhead box E1/thyroid transcription factor 2 (FOXE1/TTF-2) cause syndromic congenital hypothyroidism, with thyroid dysgenesis, cleft palate, spiky hair, and variable choanal atresia and bifid epiglottis in three cases reported hitherto. We have elucidated the molecular basis of the disorder in a female with a similar clinical phenotype, born to nonconsanguineous parents. OBJECTIVE AND DESIGN: The FOXE1 gene, located on chromosome 9q22, was sequenced in the proband and family members. Microsatellite marker and multiplex ligation probe amplification analyses determined chromosomal inheritance patterns and FOXE1 copy number. Mutant FOXE1 function was predicted by structural modeling and tested in transfection assays. RESULTS: The proband was homozygous for a novel missense (c.412T-->C; F137S) FOXE1 mutation, but her mother showed heterozygous and father wild-type alleles for this gene sequence. However, the proband was also homozygous for 10 microsatellite markers spanning chromosome 9 with exclusively maternal inheritance. Multiplex ligation probe amplification assays showed two copies of FOXE1 in the proband, indicating maternal isodisomy for chromosome 9. Consistent with structural modeling, the F137S mutant FOXE1 protein failed to bind DNA and showed negligible transcriptional activity. CONCLUSION: We have described the first case of uniparental disomy causing homozygosity for a novel, loss-of-function FOXE1/TTF-2 mutation in dysgenetic congenital hypothyroidism.


Assuntos
Cromossomos Humanos Par 9/genética , Hipotireoidismo Congênito/genética , Fatores de Transcrição Forkhead/genética , Dissomia Uniparental/genética , Adolescente , Fissura Palatina/genética , Feminino , Genótipo , Homozigoto , Humanos , Repetições de Microssatélites , Mutação
17.
J Travel Med ; 17(1): 66-8, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20074102

RESUMO

We describe seven cases of meningitis in a group of young Italian travelers coming back from India. Virologic studies identified echovirus-4 as the cause of this cluster of cases, the first imported echovirus outbreak in Italy. Enteroviruses may play an important role in undiagnosed fevers in travelers.


Assuntos
Infecções por Enterovirus/diagnóstico , Infecções por Enterovirus/epidemiologia , Meningite Viral/epidemiologia , Meningite Viral/virologia , Viagem , Adolescente , Adulto , Surtos de Doenças , Enterovirus Humano B/isolamento & purificação , Infecções por Enterovirus/transmissão , Feminino , Humanos , Índia , Itália/epidemiologia , Masculino , Meningite Viral/diagnóstico , Meningite Viral/transmissão , Reação em Cadeia da Polimerase , Adulto Jovem
19.
Travel Med Infect Dis ; 6(1-2): 32-5, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18342271

RESUMO

Early diagnosis and appropriate empirical treatment of bacterial meningitis reduce morbidity and mortality. Prevalence rates of different causative pathogens associated with bacterial meningitis can depend on age, the underlying medical condition, way of infection and geographical distribution. Klebsiella pneumoniae represents an infrequent cause of community-acquired meningitis in South-East Asia and North-East Asia, where it accounts for 20% of all bacterial meningitis, frequently associated with septic metastatic complications. We describe a case of K. pneumoniae meningitis, diplopia and chemosis in a recently immigrated patient with impaired glucose tolerance. The reason for the high prevalence of metastatic septic infections caused by K. pneumoniae in Taiwan and South-East Asia remains unclear: high prevalence in this area of serotype K1 and K2 and the expression of a novel locus called magA conferring to bacterium an elevated phagocytosis resistance and an active proliferation ability have been suggested. A high degree of suspicion for this etiology must be taken into account in immigrants from China and Taiwan. Due to a very high lethality, guidelines on empiric treatment should be considered in the management of bacterial meningitis, with the patients geographical origin and the clinical syndrome (meningitis and endophtalmitis) as potential risk factors for K. pneumoniae infection.


Assuntos
Infecções Comunitárias Adquiridas/diagnóstico , Endoftalmite/diagnóstico , Infecções por Klebsiella/diagnóstico , Klebsiella pneumoniae/patogenicidade , Meningites Bacterianas/diagnóstico , Viagem , Adulto , China/etnologia , Infecções Comunitárias Adquiridas/epidemiologia , Endoftalmite/epidemiologia , Intolerância à Glucose/complicações , Humanos , Infecções por Klebsiella/epidemiologia , Klebsiella pneumoniae/isolamento & purificação , Masculino , Meningites Bacterianas/epidemiologia , Fatores de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA